IL 15 Mouse

Interleukin-15 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to IL-15 Mouse

Interleukin-15 (IL-15) is a 14 kDa cytokine critical for immune regulation, structurally related to IL-2 but with distinct functional roles. Mouse IL-15 shares 70% amino acid sequence identity with human IL-15 and 96% with rat IL-15, making it a key model for translational studies . It is essential for the development, survival, and activation of natural killer (NK) cells, memory CD8+ T cells, and intestinal intraepithelial lymphocytes (IELs) . Unlike IL-2, IL-15 does not promote activation-induced cell death (AICD) or regulatory T cell expansion, enabling sustained immune responses .

Table 1: Sequence Identity of IL-15 Across Species

SpeciesAmino Acid Identity vs. Mouse IL-15
Human70%
Rat96%
Data sourced from R&D Systems

Transpresentation Mechanism

IL-15 primarily acts via transpresentation, where IL-15/IL-15Rα complexes on stromal or dendritic cells activate adjacent cells expressing IL-2Rβ/γc . This mechanism is critical for NK cell maturation and memory CD8+ T cell survival .

Immune Cell Development

  • NK Cells: IL-15-deficient mice lack functional NK cells, demonstrating its obligate role in NK cell ontogeny .

  • Memory CD8+ T Cells: IL-15 sustains memory CD8+ T cell populations via Bcl-xL-mediated anti-apoptotic signaling .

  • Intestinal IELs: IL-15 deficiency reduces γδ T cell and CD8αα+ IEL populations, critical for mucosal immunity .

Metabolic and Muscle Phenotypes

  • IL-15Rα Knockout (KO) Mice: Exhibit enhanced oxidative muscle fiber composition, increased fatigue resistance, and elevated mitochondrial biogenesis in fast-twitch muscles .

  • Exercise Response: IL-15 release from skeletal muscle post-exercise correlates with fat oxidation and metabolic adaptation .

Table 2: Key Phenotypes of IL-15/IL-15Rα-Deficient Mice

ModelPhenotypeMechanism
IL-15 KOAbsent NK cells, reduced memory CD8+ T cellsLoss of survival signals
IL-15Rα KOOxidative muscle shift, improved enduranceMitochondrial biogenesis
Data from Kennedy et al. (2000) and Quinn et al. (2011)

Circulating IL-15: Free vs. Complexed Forms

Contrary to earlier assumptions, 86% of serum IL-15 in mice exists in free form, while only 14% is complexed with soluble IL-15Rα (sIL-15Rα) . This challenges the traditional model of IL-15 secretion being strictly dependent on IL-15Rα and suggests alternative secretion pathways .

Cancer Immunotherapy

  • IL-15·sIL-15Rα/Fc Complex: A recombinant fusion protein with an 18-fold extended half-life (13.1 hours vs. 0.7 hours for monomeric IL-15) enhances CD8+ T cell expansion and synergizes with PD-1 blockade to inhibit tumor growth .

Table 3: Anti-Tumor Efficacy of IL-15·sIL-15Rα/Fc

ModelOutcomeCitation
HT-29 xenograftTumor growth inhibition
Combo with PD-1 AbEnhanced efficacy vs. monotherapy

Genetic Models

  • NXG hIL-15 Mouse: A humanized model expressing human IL-15 under physiological regulation (serum levels: 80 pg/mL basal, 400 pg/mL during inflammation) . Used to study human NK cell reconstitution and cytokine dynamics .

Controversies and Emerging Insights

  • IL-15 Secretion: The predominance of free IL-15 in serum suggests non-canonical secretion pathways independent of IL-15Rα, potentially involving skeletal muscle .

  • Species-Specific Effects: Human IL-15 exhibits limited cross-reactivity with mouse receptors, necessitating humanized models for translational studies .

Product Specs

Introduction
The cytokine encoded by this gene plays a crucial role in regulating the activation and proliferation of T cells and natural killer cells. This cytokine shares significant biological similarities with interleukin-2 (IL-2), including binding to common hematopoietin receptor subunits. This competition for the same receptor suggests a potential for negative regulation of each other's activity. A key observation is the influence of the balance between this cytokine and IL-2 on the number of CD8+ memory cells. Notably, this cytokine triggers the activation of JAK kinases, leading to the phosphorylation and activation of transcription activators STAT3, STAT5, and STAT6. Research on the mouse counterpart suggests that this cytokine might elevate the expression of the apoptosis inhibitor BCL2L1/BCL-x(L), potentially mediated through STAT6's transcription activation activity, thereby preventing apoptosis. Interestingly, two alternatively spliced transcript variants of this gene have been identified, both encoding the same protein.
Description
Recombinant Mouse Interleukin-15, produced in E. coli, is a single, non-glycosylated polypeptide chain. This protein consists of 152 amino acids, spanning from positions 49 to 162, and has a molecular weight of 17.6 kDa. For purification purposes, a 37 amino acid His Tag is fused to the N-terminus of IL-15, followed by purification using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The mouse IL-15 protein is supplied in a solution of phosphate-buffered saline (PBS) at a pH of 7.4, supplemented with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the protein should be kept at a refrigerated temperature of 4°C. For extended storage, it is recommended to freeze the protein at -20°C. To further enhance stability during long-term storage, adding a carrier protein like HSA or BSA (0.1%) is advisable. It's important to minimize repeated freeze-thaw cycles to maintain protein integrity.
Purity
Analysis by SDS-PAGE confirms a purity exceeding 90%.
Biological Activity
The biological activity of the protein was assessed using a cell proliferation assay with CTLL2 mouse cytotoxic T cells. The half-maximal effective concentration (ED50) was determined to be less than 1 ng/ml. This translates to a specific activity of 1,000,000 international units (IU) per milligram (mg).
Synonyms
IL-15, MGC9721, Interleukin-15, Il15, AI503618.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSHMNW IDVRYDLEKI ESLIQSIHID TTLYTDSDFH PSCKVTAMNC FLLELQVILH
EYSNMTLNET VRNVLYLANS TLSSNKNVAE SGCKECEELE EKTFTEFLQS FIRIVQMFIN TS.

Q&A

What is IL-15 and what are its main functions in the mouse immune system?

IL-15 is a pro-inflammatory cytokine critical for the development, maintenance, and response of T cells, natural killer (NK) cells, NK T cells, and dendritic cells . In the mouse immune system, IL-15 stimulates the proliferation of activated T cells, NK cells, and B cells, and induces antibody production by B cells stimulated with anti-IgM or CD40L . IL-15 also plays crucial roles in promoting the development of dendritic cells and inducing the production of proinflammatory cytokines from macrophages .

Unlike many other cytokines, IL-15 exists in two soluble forms: as a monomer (sIL-15) or as a heterodimeric complex in association with sIL-15Rα (sIL-15/IL-15Rα) . Both forms have distinct biological activities and have been tested in experimental tumor murine models. IL-15 functions are particularly important for memory CD8+ T cells, as demonstrated by studies showing that dendritic cells mediate the effects of IL-15 on these memory T cells .

Which cell types produce IL-15 in mice?

IL-15 is produced mainly by dendritic cells (DCs), epithelial cells, fibroblasts, and monocytes in mice . DCs produce IL-15 in response to type I IFN, double-stranded RNA, or lipopolysaccharide and are then activated to upregulate co-stimulatory molecules, increase production of IFN-γ, and develop an enhanced capability to stimulate T lymphocytes .

Mast cells also express both constitutive and lipopolysaccharide-inducible IL-15 and store it intracellularly . Using specialized reporter mouse lines such as the IL-15-CFP knock-in mice, researchers have identified distinct stromal cell populations that express IL-15 in primary and secondary lymphoid organs . These include fibroblastic reticular cells (FRCs) and other stromal cell subtypes that form an "IL-15 niche" with a specific distribution pattern .

How do researchers distinguish between different forms of IL-15 in mouse studies?

Distinguishing between monomeric IL-15 (sIL-15) and the heterodimeric complex (sIL-15/IL-15Rα) in mouse studies requires specific experimental approaches. Researchers typically use Western blotting with antibodies that specifically recognize either IL-15 or IL-15Rα to detect these different forms. ELISA methods have been developed to separately quantify monomeric IL-15 and the IL-15/IL-15Rα complex, though there have been controversies regarding the detection of sIL-15/IL-15Rα in plasma of healthy donors or patients with cancer .

For functional studies, recombinant versions of both forms can be used to determine their distinct biological activities. The biological significance of these different isoforms can be assessed by examining their effects on immune cell populations, particularly NK cells and memory CD8+ T cells . Research indicates that these different forms may have distinct impacts on immune responses, with prolonged exposure potentially leading to NK hyporesponsiveness through different mechanisms .

What mouse models are available for studying IL-15 function?

Several mouse models have been developed to study IL-15 function:

How does prolonged IL-15 exposure affect NK cell functionality in mice?

Prolonged exposure to IL-15 has complex effects on NK cell functionality in mice. Research suggests that in vivo prolonged or repeated exposure to monomeric sIL-15 or the soluble IL-15/IL-15Rα complex may lead to NK hypo-responsiveness through multiple mechanisms . One mechanism involves the expansion of the CD8+/CD44+ T cell subset that suppresses NK cell functions .

In vitro experiments indicate that both soluble IL-15/IL-15Rα complex and monomeric IL-15 may cause NK hyporesponsiveness through direct effects resulting from prolonged stimulation . This suggests that optimal therapeutic use of IL-15 requires careful consideration of concentrations and duration of exposure to avoid detrimental effects on NK cell function.

The development of very long-acting IL-15 formulations, such as hydrogel microspheres covalently attached to IL-15 (MS~IL-15), demonstrates that maintaining IL-15 within a narrow therapeutic window for extended periods can result in prolonged expansion of NK cells for about 2 weeks and CD44hiCD8+ T cells for about 4 weeks . This approach may help overcome the limitations of short half-life while avoiding the negative effects of excessive exposure.

What are the challenges in studying IL-15 trans-presentation in mouse models?

Studying IL-15 trans-presentation in mouse models presents several challenges:

  • Complex cellular interactions: IL-15 trans-presentation involves the presentation of IL-15 bound to IL-15Rα by one cell to another cell expressing IL-2Rβ/γc. This requires sophisticated imaging or co-culture systems to visualize and quantify these interactions.

  • Distinguishing trans-presentation from other mechanisms: It can be difficult to separate the effects of trans-presented IL-15 from those of soluble IL-15 or the soluble IL-15/IL-15Rα complex in vivo.

  • Cell-specific effects: Different cell types respond differently to trans-presented IL-15. For example, follicular dendritic cells capture IL-15 via IL-15Rα and trans-present it to promote germinal center B lymphocyte survival and proliferation, leading to the development of high-affinity antibodies .

  • Technical limitations in detection: The relatively low expression levels of IL-15 and the complex dynamics of its presentation make it challenging to track IL-15 trans-presentation events in real-time in vivo.

Researchers have addressed these challenges by developing specialized mouse models, such as the IL-15-CFP knock-in mice that allow visualization of IL-15-expressing cells , and through the use of bone marrow chimeras to delineate the roles of IL-15 expression by different cell types.

What are the optimal experimental designs for testing IL-15-based cancer immunotherapies in mouse models?

Optimal experimental designs for testing IL-15-based cancer immunotherapies in mouse models should address several key considerations:

  • Selection of appropriate tumor models: Researchers should choose tumor models that reflect the immune dependence seen in human cancers. Both NK cell-driven models (such as the MET-1 murine model of adult T-cell leukemia) and CD8+ T cell-driven models (such as the bilateral TRAMP-C2 model of prostatic cancer) have been used to evaluate IL-15-based therapies .

  • Formulation optimization: Given IL-15's short half-life, researchers have developed various approaches to extend its duration of action. For example, hydrogel microspheres covalently attached to IL-15 (MS~IL-15) by a releasable linker provide very long-acting IL-15 with low Cmax that elicits prolonged expansion of target immune cells and high anticancer activity .

  • Combination strategies: Testing IL-15 in combination with other immunotherapeutic agents often provides enhanced efficacy. For example, in the NK cell-driven MET-1 model, combining MS~IL-15 with anti-CCR4 significantly extended survival compared to either agent alone through antibody-dependent cellular cytotoxicity (ADCC) .

  • Monitoring immune responses: Comprehensive immune monitoring should include assessment of:

    • NK and CD8+ T cell expansion and activation

    • Tumor infiltration by effector cells

    • Cytokine production profiles

    • Development of immunological memory

  • Pharmacokinetic/pharmacodynamic correlation: Careful analysis of the relationship between IL-15 levels, immune cell responses, and anti-tumor effects is essential. The pharmacokinetics of MS~IL-15, for instance, showed a long half-life of about 168 hours over the first 5 days, followed by an abrupt decrease to about ~30 hours due to the development of a cytokine sink .

How can researchers engineer improved IL-15-based therapeutics using mouse models?

Engineering improved IL-15-based therapeutics using mouse models involves several strategic approaches:

  • Stability enhancement through disulfide bond engineering: Rational design of disulfide bonds has been used to improve protein stability. Researchers have employed computational modeling approaches to engineer new disulfide bonds between IL-15 and the Sushi domain of IL-15Rα based on co-crystal structures . This two-step approach involves first selecting potential mutation sites to pair disulfide bonds, then evaluating the energetic effects on protein stability for the selected residues.

  • Half-life extension strategies: Various approaches can extend IL-15's short half-life:

    • Fc fusion: A common way to improve the half-life of a molecule, successfully applied to IL-15-based molecules

    • Controlled release formulations: Hydrogel microspheres covalently attached to IL-15 (MS~IL-15) by a releasable linker can maintain IL-15 within a narrow therapeutic window for extended periods

    • Protein engineering: Introduction of inter-molecular disulfide bonds between IL-15 and its receptor α to improve developability

  • Leveraging trans-presentation mechanisms: By understanding the activity enhancement effect of the trans-presentation mechanism of IL-15 and its receptor α, researchers can design more potent molecules .

  • In vivo validation: Mouse models provide critical platforms for validating these engineered molecules. The pharmacokinetics and pharmacodynamics of engineered IL-15 should be determined in appropriate mouse strains (such as C57BL/6J), and their antitumor activity tested in relevant cancer models both as single agents and in combination with other therapeutics .

What role does IL-15 play in mouse models of viral infection?

IL-15 plays critical roles in immune responses to viral infections in mouse models:

  • Herpes simplex virus (HSV) infections: In studies of HSV-2, IL-15 signaling through the IL-2/IL-15Rβ chain is crucial for mounting protective immune responses against systemic infection . Protection against genital HSV-2 infection was critically dependent on the presence of IL-15, NK and NK T cells in a mouse model of genital herpes infection .

  • Balance of immune responses: Interestingly, despite IL-15 transgenic mice having higher numbers of NK cells in their genital mucosa and more CD8+ T lymphocytes than control mice, HSV-2 immunized IL-15 transgenic mice were susceptible to genital HSV-2 infection following challenge. This finding was attributed to a reduction in HSV-2-specific CD4+ T lymphocytes due to competition from the increased numbers of CD8+ lymphocytes produced by excess IL-15 .

  • Regulation of inflammatory responses: In some viral models, susceptibility to genital HSV-2 infection in IL-15 transgenic mice was attributed to aberrantly high levels of TGF-β1 and decreased levels of IFN-γ being produced by the intraepithelial cells, in addition to impaired production of IFN-γ by T lymphocytes .

  • Control of early viral replication: IL-15 has been shown to contribute to the control of HSV-1 (which causes mild primary infections early in life) by enhancing NK cell activity that suppresses viral replication .

These findings highlight the complex role of IL-15 in antiviral immunity, demonstrating that while IL-15 is essential for effective immune responses, excessive IL-15 can disrupt the balance of immune cell populations and impair pathogen-specific responses.

How can researchers effectively measure IL-15 protein levels in mouse tissues?

Effectively measuring IL-15 protein levels in mouse tissues requires specialized techniques due to the generally low expression levels and the complexity of IL-15 biology:

  • ELISA-based methods: While standard ELISA kits are available for mouse IL-15, they often have limitations in sensitivity and may not distinguish between different forms of IL-15 (monomeric vs. complexed with IL-15Rα). Specialized ELISAs have been developed to separately quantify monomeric IL-15 and the IL-15/IL-15Rα complex .

  • Flow cytometry: Intracellular staining for IL-15 can be performed on single-cell suspensions from various tissues, though this typically requires stimulation conditions or protein transport inhibitors to enhance detection.

  • Reporter mouse models: The IL-15-CFP knock-in mouse model provides a sensitive tool to visualize IL-15-expressing cells in vivo through fluorescence detection methods . This approach allows for identification of IL-15-expressing cells in their native tissue context.

  • Immunohistochemistry/Immunofluorescence: Tissue sections can be stained for IL-15 and IL-15Rα to visualize their expression patterns in situ, though these techniques may require signal amplification methods due to typically low expression levels.

  • Western blotting: This can be used to detect different forms of IL-15 based on molecular weight, particularly when combined with immunoprecipitation to enhance sensitivity.

When measuring IL-15 levels, researchers should be aware of age-dependent augmentation and LPS-induced enhancement of IL-15 expression in some stromal cells, as demonstrated in the IL-15-CFP knock-in mouse model .

What are the best approaches for studying IL-15-dependent immune cell development in mice?

Studying IL-15-dependent immune cell development in mice requires multiple complementary approaches:

  • Genetic models:

    • IL-15 knockout mice: Allow examination of immune cell populations that strictly depend on IL-15 for development

    • IL-15Rα knockout mice: Help distinguish receptor-dependent functions

    • IL-15 reporter mice (e.g., IL-15-CFP knock-in): Permit visualization of IL-15-expressing cells and their interactions with developing immune cells

  • Cell fate mapping: Using genetic fate-mapping approaches to track the development of IL-15-dependent cell lineages over time.

  • Bone marrow chimeras: Creating mixed bone marrow chimeras between wild-type and IL-15-deficient mice helps determine the cell-intrinsic versus cell-extrinsic requirements for IL-15 in different immune cell populations.

  • Ex vivo culture systems: Developing primary immune cells in the presence or absence of IL-15 can reveal its direct effects on cellular differentiation and function.

  • Anti-IL-15 neutralization: Administering anti-IL-15 antibodies (such as clone AIO.3) at different developmental stages can reveal critical windows of IL-15 dependence .

  • Single-cell analysis: Techniques like single-cell RNA sequencing can identify distinct developmental trajectories and gene expression programs in IL-15-dependent cell populations.

  • Cellular niches: Characterizing the specialized microenvironments supporting IL-15-dependent cell development, such as those formed by IL-15-expressing stromal cells in primary and secondary lymphoid organs .

Research has revealed that IL-15-expressing stromal cells show a distinct distribution in primary and secondary lymphoid organs and that there is age-dependent augmentation and LPS-induced enhancement of IL-15 expression in some stromal cells .

How can IL-15 function be effectively blocked in mouse models of inflammatory disease?

Effectively blocking IL-15 function in mouse models of inflammatory disease can be achieved through several approaches:

  • Neutralizing antibodies: Anti-IL-15 monoclonal antibodies, such as the AIO.3 clone, can be administered to neutralize circulating IL-15 . These antibodies bind to mouse IL-15 and prevent its interaction with receptor components, effectively blocking its biological activity.

  • Receptor blockade: Antibodies targeting IL-15Rα or IL-2/IL-15Rβ can prevent IL-15 signaling without directly neutralizing the cytokine.

  • Genetic approaches: Conditional knockout models using Cre-loxP systems can delete IL-15 or IL-15Rα in specific cell types or at specific time points to dissect their role in disease pathogenesis.

  • Soluble receptors: Recombinant soluble IL-15Rα can act as a decoy receptor, binding to IL-15 and preventing its interaction with membrane-bound receptors on target cells.

  • Small molecule inhibitors: Inhibitors targeting the downstream signaling pathways activated by IL-15, such as JAK/STAT inhibitors, can block IL-15-mediated effects.

IL-15 has been shown to play a role in several inflammatory disorders, including rheumatoid arthritis, psoriasis, and pulmonary inflammatory diseases . Blocking IL-15 in these mouse models can help elucidate its contribution to disease pathogenesis and evaluate the therapeutic potential of IL-15 pathway inhibition.

When designing studies to block IL-15 function, researchers should consider the potential compensatory mechanisms that might emerge, as well as the different effects of blocking various forms of IL-15 (monomeric vs. complexed with IL-15Rα).

Product Science Overview

Introduction

Interleukin-15 (IL-15) is a cytokine that plays a crucial role in the immune system by promoting the proliferation and maintenance of natural killer (NK) cells and CD8+ T cells. It is a member of the four α-helix bundle cytokine family and shares many biological properties with interleukin-2 (IL-2). IL-15 is produced by various cell types, including epithelial cells, monocytes, muscle cells, and placenta cells .

Structure and Function

IL-15 is a potent lymphoid cell growth factor. It stimulates the proliferation of activated T cells and promotes the generation of cytotoxic T lymphocytes (CTLs). Additionally, IL-15 induces the generation, proliferation, and activation of NK cells and supports B cell growth and immunoglobulin production. It is also essential for the maintenance of CD8+ memory T cells .

IL-15 signals through a unique receptor complex that includes the IL-15 receptor α-chain (IL-15Rα) and shares the β- and γ-chains with the IL-2 receptor. This receptor complex allows IL-15 to exert its effects on target cells .

Recombinant Mouse IL-15

Recombinant mouse IL-15 is a laboratory-produced version of the natural cytokine, optimized for use in cell culture, differentiation studies, and functional assays. It is commonly used in research to activate and expand NK and NKT cells, differentiate NK cells from purified CD34+ cells, and expand and activate T cells .

Applications in Research and Therapy

Recombinant mouse IL-15 has several applications in both research and therapeutic contexts:

  1. Activation and Expansion of Immune Cells: IL-15 is used to activate and expand NK and NKT cells, which are critical for immune responses against infections and tumors.
  2. T Cell Differentiation and Activation: IL-15 promotes the differentiation and activation of T cells, including naive CD8+ T cells and CTLs, enhancing their ability to target and eliminate infected or cancerous cells.
  3. Cancer Immunotherapy: IL-15 has shown promise as an immuno-oncology therapeutic due to its ability to stimulate the immune system. It has been used in combination with other therapeutic agents to enhance antitumor activity .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.